1
|
Liu C, Wong PY, Wang Q, Wong HY, Huang T, Cui C, Zhang N, Cheung WH, Wong RMY. Short-chain fatty acids enhance muscle mass and function through the activation of mTOR signalling pathways in sarcopenic mice. J Cachexia Sarcopenia Muscle 2024. [PMID: 39482890 DOI: 10.1002/jcsm.13573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 07/17/2024] [Accepted: 08/06/2024] [Indexed: 11/03/2024] Open
Abstract
BACKGROUND Sarcopenia is a prevalent muscle disorder in old people leading to higher fracture rate, mortality, and other adverse clinical outcomes. Evidence indicates that short-chain fatty acids (SCFAs), which are beneficial gut microbial metabolites, were reduced in old people with sarcopenia. This study aimed to determine whether the use of SCFAs as a supplement can be a therapeutic strategy of sarcopenia in a pre-clinical model. METHODS Seven-month-old pre-sarcopenic senescent accelerated mouse prone 8 (SAMP8) mice received daily SCFAs cocktail (acetate, butyrate, and propionate) for 3 months. Age-matched senescence accelerated mouse resistant 1 (SAMR1) and SAMP8 mice receiving sodium-matched drinking water were control groups. The gut microbiota composition analysis of aged mice with or without sarcopenia was conducted by 16S rDNA sequencing. Gut barrier-related proteins and lipopolysaccharide (LPS) concentration were biomarkers of gut permeability. Colon inflammation levels, circulatory SCFAs concentration, muscle quality, function, and underlying pathways were detected by cell number counting, RT-qPCR, gas chromatography-mass spectrometry, measurements of muscle wet weight and grip strength, ex vivo functional test, treadmill endurance test, transcriptomic sequencing, morphological and immunofluorescent staining, as well as western blot. To investigate the role of mTOR signalling pathways in SCFAs treatment, C2C12 myotubes were treated with rapamycin. RESULTS Aged SAMP8 mice had different microbiota composition, and lower serum butyric acid compared with SAMR1 mice (P < 0.05). SCFAs treatment reversed the increment of colon inflammation (2.8-fold lower of il-1β) and gut barrier permeability (1.7-fold lower of LPS) in SAMP8 mice. Increased muscle mass, myofibre cross-sectional area, grip strength, twitch and tetanic force were found in SCFAs-treated mice compared with control SAMP8 mice (P < 0.05). Anti-fatigue capacity (1.6-fold) and muscle glycogen (2-fold) also improved after SCFAs treatment (P < 0.05). Transcriptomic analysis showed that AMPK, insulin, and mTOR pathways were involved in SCFAs treatment (P < 0.05). Regulation of AKT/mTOR/S6K1 and AMPK/PGC1α pathways were found. SCFAs attenuated fat infiltration and improved mitochondria biogenesis of atrophic muscle. In vitro studies indicated that SCFAs inhibited FoxO3a/Atrogin1 and activated mTOR pathways to improve myotube growth (P < 0.05), and rapamycin attenuated the effect of SCFAs through the inhibition of mTOR pathways. CONCLUSIONS This study demonstrated that bacterial metabolites SCFAs could attenuate age-related muscle loss and dysfunction, and protein synthesis-related mTOR signalling pathways were involved both in vivo and in vitro.
Collapse
Affiliation(s)
- Chaoran Liu
- Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Pui Yan Wong
- Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Qianjin Wang
- Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Hei Yuet Wong
- Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Tao Huang
- Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Can Cui
- Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Ning Zhang
- Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Wing Hoi Cheung
- Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Ronald Man Yeung Wong
- Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
2
|
Maruta H, Fujii Y, Toyokawa N, Nakamura S, Yamashita H. Effects of Bifidobacterium-Fermented Milk on Obesity: Improved Lipid Metabolism through Suppression of Lipogenesis and Enhanced Muscle Metabolism. Int J Mol Sci 2024; 25:9934. [PMID: 39337421 PMCID: PMC11432277 DOI: 10.3390/ijms25189934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 09/10/2024] [Accepted: 09/12/2024] [Indexed: 09/30/2024] Open
Abstract
Obesity is a major global health concern. Studies suggest that the gut microflora may play a role in protecting against obesity. Probiotics, including lactic acid bacteria and Bifidobacterium, have garnered attention for their potential in obesity prevention. However, the effects of Bifidobacterium-fermented products on obesity have not been thoroughly elucidated. Bifidobacterium, which exists in the gut of animals, is known to enhance lipid metabolism. During fermentation, it produces acetic acid, which has been reported to improve glucose tolerance and insulin resistance, and exhibit anti-obesity and anti-diabetic effects. Functional foods have been very popular around the world, and fermented milk is a good candidate for enrichment with probiotics. In this study, we aim to evaluate the beneficial effects of milks fermented with Bifidobacterium strains on energy metabolism and obesity prevention. Three Bifidobacterium strains (Bif-15, Bif-30, and Bif-39), isolated from newborn human feces, were assessed for their acetic acid production and viability in milk. These strains were used to ferment milk. Otsuka-Long-Evans Tokushima Fatty (OLETF) rats administered Bif-15-fermented milk showed significantly lower weight gain compared to those in the water group. The phosphorylation of AMPK was increased and the expression of lipogenic genes was suppressed in the liver of rats given Bif-15-fermented milk. Additionally, gene expression related to respiratory metabolism was significantly increased in the soleus muscle of rats given Bif-15-fermented milk. These findings suggest that milk fermented with the Bifidobacterium strain Bif-15 can improve lipid metabolism and suppress obesity.
Collapse
Affiliation(s)
- Hitomi Maruta
- Department of Nutritional Science, Faculty of Health and Welfare Science, Okayama Prefectural University, 111 Kuboki, Soja-shi 719-1197, Okayama, Japan
| | - Yusuke Fujii
- Fundamental Laboratory, Ohayo Daily Products Co., Ltd., 565 Koshita, Naka-ku, Okayama-shi 703-8505, Okayama, Japan
| | - Naoki Toyokawa
- Fundamental Laboratory, Ohayo Daily Products Co., Ltd., 565 Koshita, Naka-ku, Okayama-shi 703-8505, Okayama, Japan
| | - Shoji Nakamura
- Fundamental Laboratory, Ohayo Daily Products Co., Ltd., 565 Koshita, Naka-ku, Okayama-shi 703-8505, Okayama, Japan
| | - Hiromi Yamashita
- Department of Nutritional Science, Faculty of Health and Welfare Science, Okayama Prefectural University, 111 Kuboki, Soja-shi 719-1197, Okayama, Japan
- Graduate School of Health and Welfare Science, Okayama Prefectural University, 111 Kuboki, Soja-shi 719-1197, Okayama, Japan
| |
Collapse
|
3
|
Adiamo OQ, Bobasa EM, Phan ADT, Akter S, Seididamyeh M, Dayananda B, Gaisawat MB, Kubow S, Sivakumar D, Sultanbawa Y. In-vitro colonic fermentation of Kakadu plum (Terminalia ferdinandiana) fruit powder: Microbial biotransformation of phenolic compounds and cytotoxicity. Food Chem 2024; 448:139057. [PMID: 38555694 DOI: 10.1016/j.foodchem.2024.139057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 01/26/2024] [Accepted: 03/15/2024] [Indexed: 04/02/2024]
Abstract
Kakadu plum (Terminalia ferdinandiana) (KP) is an indigenous fruit used as a functional ingredient in powdered form. Three KP doses (1, 2.5 and 5 g) were digested in a dynamic in vitro gut digestion model over 48 h. Faecal water digests from the colonic reactors were assessed for total soluble polyphenols (TSP), ferric reducing antioxidant power (FRAP), phenolic metabolites and short-chain fatty acids (SCFAs). Effects of digests on cell viability were tested against Caco-2 intestinal and HepG2 hepatic cells. All doses of KP fermentation produced castalagin, corilagin, chebulagic acid, chebulinic acid, and gallic acid. TSP and FRAP significantly increased in 5 g KP digests at 0 and 48 h of fermentation. SCFA concentrations significantly increased after 48 h. Cytotoxic effects of 2.5 and 5 g KP digests diminished significantly after 12 h. Overall, colonic fermentation increased antioxidant activity and polyphenolic metabolites of 5 g KP powder for 48 h.
Collapse
Affiliation(s)
- Oladipupo Q Adiamo
- ARC Industrial Transformation Training Centre for Uniquely Australian Foods, Queensland Alliance for Agriculture and Food Innovation (QAAFI), The University of Queensland (UQ), Indooroopilly, QLD 4068, Australia
| | - Eshetu M Bobasa
- ARC Industrial Transformation Training Centre for Uniquely Australian Foods, Queensland Alliance for Agriculture and Food Innovation (QAAFI), The University of Queensland (UQ), Indooroopilly, QLD 4068, Australia
| | - Anh Dao Thi Phan
- ARC Industrial Transformation Training Centre for Uniquely Australian Foods, Queensland Alliance for Agriculture and Food Innovation (QAAFI), The University of Queensland (UQ), Indooroopilly, QLD 4068, Australia
| | - Saleha Akter
- ARC Industrial Transformation Training Centre for Uniquely Australian Foods, Queensland Alliance for Agriculture and Food Innovation (QAAFI), The University of Queensland (UQ), Indooroopilly, QLD 4068, Australia
| | - Maral Seididamyeh
- ARC Industrial Transformation Training Centre for Uniquely Australian Foods, Queensland Alliance for Agriculture and Food Innovation (QAAFI), The University of Queensland (UQ), Indooroopilly, QLD 4068, Australia
| | - Buddhi Dayananda
- School of Agriculture and Food Sciences, The University of Queensland, Brisbane, QLD 4072, Australia
| | | | - Stan Kubow
- School of Human Nutrition, McGill University, Montréal, QC, Canada
| | - Dharini Sivakumar
- ARC Industrial Transformation Training Centre for Uniquely Australian Foods, Queensland Alliance for Agriculture and Food Innovation (QAAFI), The University of Queensland (UQ), Indooroopilly, QLD 4068, Australia; Phytochemical Food Network, Department of Crop Sciences, Tshwane University of Technology, Pretoria 001, South Africa
| | - Yasmina Sultanbawa
- ARC Industrial Transformation Training Centre for Uniquely Australian Foods, Queensland Alliance for Agriculture and Food Innovation (QAAFI), The University of Queensland (UQ), Indooroopilly, QLD 4068, Australia.
| |
Collapse
|
4
|
Tang MY, Xie H, Tao JT, Zhang C, Luo YH, Zhang C, Peng SQ, Xie LX, Lv WB, Zhang C, Huang L. Pathophysiological relevance and therapeutic outlook of GPR43 in atherosclerosis. Biochem Cell Biol 2024. [PMID: 39013204 DOI: 10.1139/bcb-2024-0053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/18/2024] Open
Abstract
Atherosclerosis (AS) is an inflammatory arterial disorder that occurs due to the deposition of the excessive lipoprotein under the artery intima, mainly including low-density lipoprotein and other apolipoprotein B-containing lipoproteins. G protein-coupled receptors (GPCRs) play a crucial role in transmitting signals in physiological and pathophysiological conditions. GPCRs recognize inflammatory mediators, thereby serving as important players during chronic inflammatory processes. It has been demonstrated that free fatty acids can function as ligands for various GPCRs, such as free fatty acid receptor (FFAR)1/GPR40, FFAR2/GPR43, FFAR3/GPR41, FFAR4/GPR120, and the lipid metabolite binding glucose-dependent insulinotropic receptor (GPR119). This review discusses GPR43 and its ligands in the pathogenesis of AS, especially focusing on its distinct role in regulating chronic vascular inflammation, inhibiting oxidative stress, ameliorating endothelial dysfunction and improving dyslipidemia. It is hoped that this review may provide guidance for further studies aimed at GPR43 as a promising target for drug development in the prevention and therapy of AS.
Collapse
Affiliation(s)
- Mu-Yao Tang
- Research Laboratory of Translational Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, People's Republic of China
- Departments of Clinical Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, People's Republic of China
| | - Hao Xie
- Research Laboratory of Translational Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, People's Republic of China
- Departments of Clinical Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, People's Republic of China
| | - Jin-Tao Tao
- Research Laboratory of Translational Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, People's Republic of China
- Departments of Clinical Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, People's Republic of China
| | - Chun Zhang
- Research Laboratory of Translational Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, People's Republic of China
- Departments of Clinical Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, People's Republic of China
| | - Yao-Hua Luo
- Research Laboratory of Translational Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, People's Republic of China
- Departments of Clinical Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, People's Republic of China
| | - Cong Zhang
- Research Laboratory of Translational Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, People's Republic of China
- Departments of Clinical Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, People's Republic of China
| | - Si-Qin Peng
- Research Laboratory of Translational Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, People's Republic of China
- Departments of Clinical Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, People's Republic of China
| | - Lin-Xi Xie
- Research Laboratory of Translational Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, People's Republic of China
- Departments of Clinical Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, People's Republic of China
| | - Wen-Bo Lv
- Research Laboratory of Translational Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, People's Republic of China
- Departments of Clinical Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, People's Republic of China
| | - Chi Zhang
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, People's Republic of China
| | - Liang Huang
- Research Laboratory of Translational Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, People's Republic of China
| |
Collapse
|
5
|
Lee B, Lee SM, Song JW, Choi JW. Gut Microbiota Metabolite Messengers in Brain Function and Pathology at a View of Cell Type-Based Receptor and Enzyme Reaction. Biomol Ther (Seoul) 2024; 32:403-423. [PMID: 38898687 PMCID: PMC11214962 DOI: 10.4062/biomolther.2024.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 05/02/2024] [Accepted: 05/25/2024] [Indexed: 06/21/2024] Open
Abstract
The human gastrointestinal (GI) tract houses a diverse microbial community, known as the gut microbiome comprising bacteria, viruses, fungi, and protozoa. The gut microbiome plays a crucial role in maintaining the body's equilibrium and has recently been discovered to influence the functioning of the central nervous system (CNS). The communication between the nervous system and the GI tract occurs through a two-way network called the gut-brain axis. The nervous system and the GI tract can modulate each other through activated neuronal cells, the immune system, and metabolites produced by the gut microbiome. Extensive research both in preclinical and clinical realms, has highlighted the complex relationship between the gut and diseases associated with the CNS, such as Alzheimer's disease, Parkinson's disease, and amyotrophic lateral sclerosis. This review aims to delineate receptor and target enzymes linked with gut microbiota metabolites and explore their specific roles within the brain, particularly their impact on CNS-related diseases.
Collapse
Affiliation(s)
- Bada Lee
- Department of Biomedicinal and Pharmaceutical Sciences, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Soo Min Lee
- Department of Biomedicinal and Pharmaceutical Sciences, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Jae Won Song
- Department of Regulatory Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Jin Woo Choi
- Department of Biomedicinal and Pharmaceutical Sciences, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Regulatory Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| |
Collapse
|
6
|
Lee DH, Kim MT, Han JH. GPR41 and GPR43: From development to metabolic regulation. Biomed Pharmacother 2024; 175:116735. [PMID: 38744220 DOI: 10.1016/j.biopha.2024.116735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 05/06/2024] [Accepted: 05/07/2024] [Indexed: 05/16/2024] Open
Abstract
G-protein-coupled receptors are a diverse class of cell surface receptors that orchestrate numerous physiological functions. The G-protein-coupled receptors, GPR41 and GPR43, sense short-chain fatty acids (SCFAs), which are metabolites of dietary fermentation by the host's intestinal bacteria. These receptors have gained attention as potential therapeutic targets against various diseases because of their SCFA-mediated beneficial effects on the host's intestinal health. Mounting evidence has associated the activity of these receptors with chronic metabolic diseases, including obesity, diabetes, inflammation, and cardiovascular disease. However, despite intensive research using various strategies, including gene knockout (KO) mouse models, evidence about the precise roles of GPR41 and GPR43 in disease treatment remains inconsistent. Here, we comprehensively review the latest findings from functional studies of the signaling mechanisms that underlie the activities of GPR41 and GPR43, as well as highlight their multifaceted roles in health and disease. We anticipate that this knowledge will guide future research priorities and the development of effective therapeutic interventions.
Collapse
Affiliation(s)
- Do-Hyung Lee
- College of Pharmacy, Chungnam National University, Daejeon 34134, the Republic of Korea
| | - Min-Tae Kim
- Department of Pharmaceutical Research, KyongBo Pharmaceutical Co., Ltd, 174, Sirok-ro, Asan-si, Chungcheongnam-do 31501, the Republic of Korea
| | - Joo-Hui Han
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Woosuk University, Wanju 55338, the Republic of Korea.
| |
Collapse
|
7
|
Ren J, Dai J, Chen Y, Wang Z, Sha R, Mao J. Physiochemical characterization and ameliorative effect of rice resistant starch modified by heat-stable α-amylase and glucoamylase on the gut microbial community in T2DM mice. Food Funct 2024; 15:5596-5612. [PMID: 38722000 DOI: 10.1039/d3fo05456j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/21/2024]
Abstract
In the presented study, natural rice containing high resistant starch content was used as a raw material to produce rice resistant starch (RRS) through enzymatic hydrolysis with heat-stable α-amylase and glucoamylase. The chemical composition, structural characteristics and in vitro glycemic index (GI) of RRS were evaluated. The effects of RRS at different doses on the body weight, serum biochemical levels, pathological indexes, production of short-chain fatty acids (SCFAs) in the gut and the intestinal microbial composition in T2DM mice were investigated. The results of physiochemical characterization indicated that, relative to rice flour, RRS mainly comprising resistant starch had higher crystallinity (25.85%) and a more stable structure, which contributed to its lower digestibility and decreased GI in vitro. Compared with the model control group, 1 g per kg BW and 2 g per kg BW oral gavage dosages of RRS effectively enhanced the SCFA productivity in the T2DM mouse gut, as well as alleviating T2DM symptoms, involving an increase in body weight, reduction in fasting blood glucose, total cholesterol, triglyceride, low-density lipoprotein cholesterol, alanine transaminase and aspartate aminotransferase, and an increase in serum insulin and high-density lipoprotein cholesterol. Besides, 1 g per kg BW and 2 g per kg BW dosages of RRS mitigated T2DM-induced pancreas damage. Furthermore, up-regulation in the abundance of probiotics (Lactobacillus, Ruminococcus, etc.) and down-regulation in the number of harmful bacteria (Desulfovibrio, Prevotella, etc.) were observed in all RRS-treated groups. In summary, this work suggested that RRS prepared using heat-stable α-amylase and glucoamylase could be a potential functional component for amelioration of T2DM applied in the fields of food and pharmaceutics.
Collapse
Affiliation(s)
- Jianing Ren
- School of Biological and Chemical Engineering, Zhejiang University of Science and Technology, Hangzhou 310023, China.
| | - Jing Dai
- School of Biological and Chemical Engineering, Zhejiang University of Science and Technology, Hangzhou 310023, China.
| | - Yue Chen
- School of Biological and Chemical Engineering, Zhejiang University of Science and Technology, Hangzhou 310023, China.
| | - Zhenzhen Wang
- School of Biological and Chemical Engineering, Zhejiang University of Science and Technology, Hangzhou 310023, China.
| | - Ruyi Sha
- School of Biological and Chemical Engineering, Zhejiang University of Science and Technology, Hangzhou 310023, China.
| | - Jianwei Mao
- School of Biological and Chemical Engineering, Zhejiang University of Science and Technology, Hangzhou 310023, China.
| |
Collapse
|
8
|
Van K, Burns JL, Monk JM. Effect of Short-Chain Fatty Acids on Inflammatory and Metabolic Function in an Obese Skeletal Muscle Cell Culture Model. Nutrients 2024; 16:500. [PMID: 38398822 PMCID: PMC10891728 DOI: 10.3390/nu16040500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 01/29/2024] [Accepted: 02/01/2024] [Indexed: 02/25/2024] Open
Abstract
The fermentation of non-digestible carbohydrates produces short-chain fatty acids (SCFAs), which have been shown to impact both skeletal muscle metabolic and inflammatory function; however, their effects within the obese skeletal muscle microenvironment are unknown. In this study, we developed a skeletal muscle in vitro model to mimic the critical features of the obese skeletal muscle microenvironment using L6 myotubes co-treated with 10 ng/mL lipopolysaccharide (LPS) and 500 µM palmitic acid (PA) for 24 h ± individual SCFAs, namely acetate, propionate and butyrate at 0.5 mM and 2.5 mM. At the lower SCFA concentration (0.5 mM), all three SCFA reduced the secreted protein level of RANTES, and only butyrate reduced IL-6 protein secretion and the intracellular protein levels of activated (i.e., ratio of phosphorylated-total) NFκB p65 and STAT3 (p < 0.05). Conversely, at the higher SCFA concentration (2.5 mM), individual SCFAs exerted different effects on inflammatory mediator secretion. Specifically, butyrate reduced IL-6, MCP-1 and RANTES secretion, propionate reduced IL-6 and RANTES, and acetate only reduced RANTES secretion (p < 0.05). All three SCFAs reduced intracellular protein levels of activated NFκB p65 and STAT3 (p < 0.05). Importantly, only the 2.5 mM SCFA concentration resulted in all three SCFAs increasing insulin-stimulated glucose uptake compared to control L6 myotube cultures (p < 0.05). Therefore, SCFAs exert differential effects on inflammatory mediator secretion in a cell culture model, recapitulating the obese skeletal muscle microenvironment; however, all three SCFAs exerted a beneficial metabolic effect only at a higher concentration via increasing insulin-stimulated glucose uptake, collectively exerting differing degrees of a beneficial effect on obesity-associated skeletal muscle dysfunction.
Collapse
Affiliation(s)
- Kelsey Van
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, ON N1G 2W1, Canada;
| | - Jessie L. Burns
- Department of Health Sciences, Carleton University, Ottawa, ON K1S 5B6, Canada;
| | - Jennifer M. Monk
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, ON N1G 2W1, Canada;
| |
Collapse
|
9
|
Durand G, Charrier P, Bes S, Bernard L, Lamothe V, Gruffat D, Bonnet M. Gene expression of free fatty acids-sensing G protein-coupled receptors in beef cattle. J Anim Sci 2024; 102:skae114. [PMID: 38659415 DOI: 10.1093/jas/skae114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 04/24/2024] [Indexed: 04/26/2024] Open
Abstract
Many physiological functions are regulated by free fatty acids (FFA). Recently, the discovery of FFA-specific G protein-coupled receptors (FFARs) has added to the complexity of their actions at the cellular level. The study of FFAR in cattle is still in its earliest stages focusing mainly on dairy cows. In this study, we set out to map the expression of genes encoding FFARs in 6 tissues of beef cattle. We also investigated the potential effect of dietary forage nature on FFAR gene expression. To this end, 16 purebred Charolais bulls were fed a grass silage ration or a maize silage ration (n = 8/group) with a forage/concentrate ratio close to 60:40 for 196 d. The animals were then slaughtered at 485 ± 42 d and liver, spleen, ileum, rectum, perirenal adipose tissue (PRAT), and Longissimus Thoracis muscle were collected. FFAR gene expression was determined by real-time quantitative PCR. Our results showed that of the five FFARs investigated, FFAR1, FFAR2, FFAR3, and GPR84 are expressed (Ct < 30) in all six tissues, whereas FFAR4 was only expressed (Ct < 30) in PRAT, ileum, and rectum. In addition, our results showed that the nature of the forage, i.e., grass silage or maize silage, had no effect on the relative abundance of FFAR in any of the tissues studied (P value > 0.05). Taken together, these results open new perspectives for studying the physiological role of these receptors in beef cattle, particularly in nutrient partitioning during growth.
Collapse
Affiliation(s)
- Guillaume Durand
- INRAE, Université Clermont Auvergne, VetagroSup, UMRH, 63122 Saint-Genès-Champanelle, France
- Bordeaux Sciences Agro, 33170 Gradignan, France
| | | | - Sébastien Bes
- INRAE, Université Clermont Auvergne, VetagroSup, UMRH, 63122 Saint-Genès-Champanelle, France
| | - Laurence Bernard
- INRAE, Université Clermont Auvergne, VetagroSup, UMRH, 63122 Saint-Genès-Champanelle, France
| | | | - Dominique Gruffat
- INRAE, Université Clermont Auvergne, VetagroSup, UMRH, 63122 Saint-Genès-Champanelle, France
| | - Muriel Bonnet
- INRAE, Université Clermont Auvergne, VetagroSup, UMRH, 63122 Saint-Genès-Champanelle, France
| |
Collapse
|
10
|
Robberechts R, Poffé C. Defining ketone supplementation: the evolving evidence for postexercise ketone supplementation to improve recovery and adaptation to exercise. Am J Physiol Cell Physiol 2024; 326:C143-C160. [PMID: 37982172 DOI: 10.1152/ajpcell.00485.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 11/14/2023] [Accepted: 11/14/2023] [Indexed: 11/21/2023]
Abstract
Over the last decade, there has been a growing interest in the use of ketone supplements to improve athletic performance. These ketone supplements transiently elevate the concentrations of the ketone bodies acetoacetate (AcAc) and d-β-hydroxybutyrate (βHB) in the circulation. Early studies showed that ketone bodies can improve energetic efficiency in striated muscle compared with glucose oxidation and induce a glycogen-sparing effect during exercise. As such, most research has focused on the potential of ketone supplementation to improve athletic performance via ingestion of ketones immediately before or during exercise. However, subsequent studies generally observed no performance improvement, and particularly not under conditions that are relevant for most athletes. However, more and more studies are reporting beneficial effects when ketones are ingested after exercise. As such, the real potential of ketone supplementation may rather be in their ability to enhance postexercise recovery and training adaptations. For instance, recent studies observed that postexercise ketone supplementation (PEKS) blunts the development of overtraining symptoms, and improves sleep, muscle anabolic signaling, circulating erythropoietin levels, and skeletal muscle angiogenesis. In this review, we provide an overview of the current state-of-the-art about the impact of PEKS on aspects of exercise recovery and training adaptation, which is not only relevant for athletes but also in multiple clinical conditions. In addition, we highlight the underlying mechanisms by which PEKS may improve exercise recovery and training adaptation. This includes epigenetic effects, signaling via receptors, modulation of neurotransmitters, energy metabolism, and oxidative and anti-inflammatory pathways.
Collapse
Affiliation(s)
- Ruben Robberechts
- Exercise Physiology Research Group, Department of Movement Sciences, KU Leuven, Leuven, Belgium
| | - Chiel Poffé
- Exercise Physiology Research Group, Department of Movement Sciences, KU Leuven, Leuven, Belgium
| |
Collapse
|
11
|
Wen C, Wang Q, Gu S, Jin J, Yang N. Emerging perspectives in the gut-muscle axis: The gut microbiota and its metabolites as important modulators of meat quality. Microb Biotechnol 2024; 17:e14361. [PMID: 37902307 PMCID: PMC10832551 DOI: 10.1111/1751-7915.14361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 09/30/2023] [Accepted: 10/11/2023] [Indexed: 10/31/2023] Open
Abstract
Animal breeding has made great genetic progress in increasing carcass weight and meat yield in recent decades. However, these improvements have come at the expense of meat quality. As the demand for meat quantity continues to rise, the meat industry faces the great challenge of maintaining and even increasing product quality. Recent research, including traditional statistical analyses and gut microbiota regulation research, has demonstrated that the gut microbiome exerts a considerable effect on meat quality, which has become increasingly intriguing in farm animals. Microbial metabolites play crucial roles as substrates or signalling factors to distant organs, influencing meat quality either beneficially or detrimentally. Interventions targeting the gut microbiota exhibit excellent potential as natural ways to foster the conversion of myofibres and promote intramuscular fat deposition. Here, we highlight the emerging roles of the gut microbiota in various dimensions of meat quality. We focus particularly on the effects of the gut microbiota and gut-derived molecules on muscle fibre metabolism and intramuscular fat deposition and attempt to summarize the potential underlying mechanisms.
Collapse
Affiliation(s)
- Chaoliang Wen
- State Key Laboratory of Animal Biotech Breeding and Frontier Science Center for Molecular Design BreedingChina Agricultural UniversityBeijingChina
- National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural AffairsChina Agricultural UniversityBeijingChina
- Department of Animal Genetics and Breeding, College of Animal Science and TechnologyChina Agricultural UniversityBeijingChina
- Sanya Institute of China Agricultural UniversityHainanChina
| | - Qunpu Wang
- State Key Laboratory of Animal Biotech Breeding and Frontier Science Center for Molecular Design BreedingChina Agricultural UniversityBeijingChina
- National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural AffairsChina Agricultural UniversityBeijingChina
- Department of Animal Genetics and Breeding, College of Animal Science and TechnologyChina Agricultural UniversityBeijingChina
| | - Shuang Gu
- State Key Laboratory of Animal Biotech Breeding and Frontier Science Center for Molecular Design BreedingChina Agricultural UniversityBeijingChina
- National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural AffairsChina Agricultural UniversityBeijingChina
- Department of Animal Genetics and Breeding, College of Animal Science and TechnologyChina Agricultural UniversityBeijingChina
| | - Jiaming Jin
- State Key Laboratory of Animal Biotech Breeding and Frontier Science Center for Molecular Design BreedingChina Agricultural UniversityBeijingChina
- National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural AffairsChina Agricultural UniversityBeijingChina
- Department of Animal Genetics and Breeding, College of Animal Science and TechnologyChina Agricultural UniversityBeijingChina
| | - Ning Yang
- State Key Laboratory of Animal Biotech Breeding and Frontier Science Center for Molecular Design BreedingChina Agricultural UniversityBeijingChina
- National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural AffairsChina Agricultural UniversityBeijingChina
- Department of Animal Genetics and Breeding, College of Animal Science and TechnologyChina Agricultural UniversityBeijingChina
- Sanya Institute of China Agricultural UniversityHainanChina
| |
Collapse
|
12
|
Wang G, Liu J, Zhang Y, Xie J, Chen S, Shi Y, Shi F, Zhu SJ. Ginsenoside Rg3 enriches SCFA-producing commensal bacteria to confer protection against enteric viral infection via the cGAS-STING-type I IFN axis. THE ISME JOURNAL 2023; 17:2426-2440. [PMID: 37950067 PMCID: PMC10689736 DOI: 10.1038/s41396-023-01541-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 10/07/2023] [Accepted: 10/10/2023] [Indexed: 11/12/2023]
Abstract
The microbiota-associated factors that influence host susceptibility and immunity to enteric viral infections remain poorly defined. We identified that the herbal monomer ginsenoside Rg3 (Rg3) can shape the gut microbiota composition, enriching robust short-chain fatty acid (SCFA)-producing Blautia spp. Colonization by representative Blautia coccoides and Blautia obeum could protect germ-free or vancomycin (Van)-treated mice from enteric virus infection, inducing type I interferon (IFN-I) responses in macrophages via the MAVS-IRF3-IFNAR signaling pathway. Application of exogenous SCFAs (acetate/propionate) reproduced the protective effect of Rg3 and Blautia spp. in Van-treated mice, enhancing intracellular Ca2+- and MAVS-dependent mtDNA release and activating the cGAS-STING-IFN-I axis by stimulating GPR43 signaling in macrophages. Our findings demonstrate that macrophage sensing of metabolites from specific commensal bacteria can prime the IFN-I signaling that is required for antiviral functions.
Collapse
Affiliation(s)
- Gan Wang
- Department of Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou, 310058, Zhejiang, PR China
| | - Jingtianyi Liu
- Department of Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou, 310058, Zhejiang, PR China
| | - Yanan Zhang
- Department of Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou, 310058, Zhejiang, PR China
| | - Jinyan Xie
- Department of Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou, 310058, Zhejiang, PR China
| | - Shuxian Chen
- Department of Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou, 310058, Zhejiang, PR China
| | - Yuhua Shi
- Department of Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou, 310058, Zhejiang, PR China
| | - Fushan Shi
- Department of Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou, 310058, Zhejiang, PR China
| | - Shu Jeffrey Zhu
- Department of Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou, 310058, Zhejiang, PR China.
| |
Collapse
|
13
|
de Souza PB, de Araujo Borba L, Castro de Jesus L, Valverde AP, Gil-Mohapel J, Rodrigues ALS. Major Depressive Disorder and Gut Microbiota: Role of Physical Exercise. Int J Mol Sci 2023; 24:16870. [PMID: 38069198 PMCID: PMC10706777 DOI: 10.3390/ijms242316870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Revised: 11/24/2023] [Accepted: 11/25/2023] [Indexed: 12/18/2023] Open
Abstract
Major depressive disorder (MDD) has a high prevalence and is a major contributor to the global burden of disease. This psychiatric disorder results from a complex interaction between environmental and genetic factors. In recent years, the role of the gut microbiota in brain health has received particular attention, and compelling evidence has shown that patients suffering from depression have gut dysbiosis. Several studies have reported that gut dysbiosis-induced inflammation may cause and/or contribute to the development of depression through dysregulation of the gut-brain axis. Indeed, as a consequence of gut dysbiosis, neuroinflammatory alterations caused by microglial activation together with impairments in neuroplasticity may contribute to the development of depressive symptoms. The modulation of the gut microbiota has been recognized as a potential therapeutic strategy for the management of MMD. In this regard, physical exercise has been shown to positively change microbiota composition and diversity, and this can underlie, at least in part, its antidepressant effects. Given this, the present review will explore the relationship between physical exercise, gut microbiota and depression, with an emphasis on the potential of physical exercise as a non-invasive strategy for modulating the gut microbiota and, through this, regulating the gut-brain axis and alleviating MDD-related symptoms.
Collapse
Affiliation(s)
- Pedro Borges de Souza
- Center of Biological Sciences, Department of Biochemistry, Universidade Federal de Santa Catarina, Florianópolis 88037-000, SC, Brazil; (P.B.d.S.); (L.d.A.B.); (L.C.d.J.); (A.P.V.)
| | - Laura de Araujo Borba
- Center of Biological Sciences, Department of Biochemistry, Universidade Federal de Santa Catarina, Florianópolis 88037-000, SC, Brazil; (P.B.d.S.); (L.d.A.B.); (L.C.d.J.); (A.P.V.)
| | - Louise Castro de Jesus
- Center of Biological Sciences, Department of Biochemistry, Universidade Federal de Santa Catarina, Florianópolis 88037-000, SC, Brazil; (P.B.d.S.); (L.d.A.B.); (L.C.d.J.); (A.P.V.)
| | - Ana Paula Valverde
- Center of Biological Sciences, Department of Biochemistry, Universidade Federal de Santa Catarina, Florianópolis 88037-000, SC, Brazil; (P.B.d.S.); (L.d.A.B.); (L.C.d.J.); (A.P.V.)
| | - Joana Gil-Mohapel
- Island Medical Program, Faculty of Medicine, University of British Columbia, Victoria, BC V8P 5C2, Canada
- Division of Medical Sciences, University of Victoria, Victoria, BC V8P 5C2, Canada
| | - Ana Lúcia S. Rodrigues
- Center of Biological Sciences, Department of Biochemistry, Universidade Federal de Santa Catarina, Florianópolis 88037-000, SC, Brazil; (P.B.d.S.); (L.d.A.B.); (L.C.d.J.); (A.P.V.)
| |
Collapse
|
14
|
Yang Y, Liu C, Zhang C, Xu Z, Zhang L, Cui Y, Wang C, Lin Y, Hou X. Acetate Upregulates GPR43 Expression and Function via PI3K-AKT-SP1 Signaling in Mammary Epithelial Cells during Milk Fat Synthesis. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:16003-16015. [PMID: 37870996 DOI: 10.1021/acs.jafc.3c00965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
This study investigated the mechanism underlying acetate-induced orphan G-protein-coupled receptor 43 (GPR43) expression and milk fat production. The mammary epithelial cells of dairy cows were treated with acetate, and the effects of GPR43 on acetate uptake and the expression of lipogenesis-related genes were determined by gas chromatography and quantitative polymerase chain reaction (qPCR), respectively. RNAi, inhibitor treatment, and luciferase assay were used to determine the effect of phosphoinositide 3-kinase-protein kinase B-specificity protein 1 (PI3K-AKT-SP1) signaling on acetate-induced GPR43 expression and function. The results showed that GPR43 was highly expressed in lactating cow mammary tissues, which was related to milk fat synthesis. 12 mM acetate significantly increased the GPR43 expression in mammary epithelial cells of dairy cows. In acetate-treated cells, GPR43 overexpression significantly increased the cellular uptake of acetate, the intracellular triacylglycerol (TAG) content, and acetate-induced lipogenesis gene expression. Acetate activated PI3K-AKT signaling and promoted SP1 translocation from the cytosol into the nucleus, where SP1 bound to the GPR43 promoter and upregulated GPR43 transcription. Moreover, the activation of PI3K-AKT-SP1 by acetate facilitated the trafficking of GPR43 from the cytosol to the plasma membrane. In conclusion, acetate upregulated GPR43 expression and function via PI3K-AKT-SP1 signaling in mammary epithelial cells, thereby increasing milk fat synthesis. These results provide an experimental strategy for improving milk lipid synthesis, which is important to the dairy industry.
Collapse
Affiliation(s)
- Yang Yang
- Key Laboratory of Animal Cellular and Genetics Engineering of Heilongjiang Province, Northeast Agricultural University, Harbin 150030, China
| | - Chuanping Liu
- Key Laboratory of Animal Cellular and Genetics Engineering of Heilongjiang Province, Northeast Agricultural University, Harbin 150030, China
| | - Caiyan Zhang
- Key Laboratory of Animal Cellular and Genetics Engineering of Heilongjiang Province, Northeast Agricultural University, Harbin 150030, China
| | - Ziru Xu
- Key Laboratory of Animal Cellular and Genetics Engineering of Heilongjiang Province, Northeast Agricultural University, Harbin 150030, China
| | - Li Zhang
- Key Laboratory of Dairy Science of Education Ministry, Northeast Agricultural University, Harbin 150030, China
| | - Yingjun Cui
- Key Laboratory of Dairy Science of Education Ministry, Northeast Agricultural University, Harbin 150030, China
| | - Chunmei Wang
- Key Laboratory of Dairy Science of Education Ministry, Northeast Agricultural University, Harbin 150030, China
| | - Ye Lin
- Key Laboratory of Animal Cellular and Genetics Engineering of Heilongjiang Province, Northeast Agricultural University, Harbin 150030, China
- Key Laboratory of Dairy Science of Education Ministry, Northeast Agricultural University, Harbin 150030, China
| | - Xiaoming Hou
- Key Laboratory of Animal Cellular and Genetics Engineering of Heilongjiang Province, Northeast Agricultural University, Harbin 150030, China
| |
Collapse
|
15
|
Zheng H, Sun Y, Zeng Y, Zheng T, Jia F, Xu P, Xu Y, Cao Y, He K, Yang Y. Effects of Four Extraction Methods on Structure and In Vitro Fermentation Characteristics of Soluble Dietary Fiber from Rape Bee Pollen. Molecules 2023; 28:4800. [PMID: 37375355 DOI: 10.3390/molecules28124800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 06/12/2023] [Accepted: 06/14/2023] [Indexed: 06/29/2023] Open
Abstract
In this study, soluble dietary fibers (SDFs) were extracted from rape bee pollen using four methods including acid extraction (AC), alkali extraction (AL), cellulase extraction (CL) and complex enzyme extraction (CE). The effects of different extraction methods on the structure of SDFs and in vitro fermentation characteristics were further investigated. The results showed that the four extraction methods significantly affected the monosaccharide composition molar ratio, molecular weight, surface microstructure and phenolic compounds content, but showed little effect on the typical functional groups and crystal structure. In addition, all SDFs decreased the Firmicutes/Bacteroidota ratio, promoted the growth of beneficial bacteria such as Bacteroides, Parabacteroides and Phascolarctobacterium, inhibited the growth of pathogenic bacteria such as Escherichia-Shigella, and increased the total short-chain fatty acids (SCFAs) concentrations by 1.63-2.45 times, suggesting that the bee pollen SDFs had a positive regulation on gut microbiota. Notably, the SDF obtained by CE exhibited the largest molecular weight, a relatively loose structure, higher extraction yield and phenolic compounds content and the highest SCFA concentration. Overall, our results indicated that CE was an appropriate extraction method of high-quality bee pollen SDF.
Collapse
Affiliation(s)
- Hui Zheng
- College of Pharmacy, Hunan University of Chinese Medicine, Changsha 410000, China
| | - Yan Sun
- College of Pharmacy, Hunan University of Chinese Medicine, Changsha 410000, China
| | - Yiqiong Zeng
- College of Pharmacy, Hunan University of Chinese Medicine, Changsha 410000, China
| | - Tao Zheng
- College of Pharmacy, Hunan University of Chinese Medicine, Changsha 410000, China
| | - Fan Jia
- College of Pharmacy, Hunan University of Chinese Medicine, Changsha 410000, China
| | - Pan Xu
- College of Pharmacy, Hunan University of Chinese Medicine, Changsha 410000, China
| | - Yao Xu
- College of Pharmacy, Hunan University of Chinese Medicine, Changsha 410000, China
| | - Yuxin Cao
- College of Pharmacy, Hunan University of Chinese Medicine, Changsha 410000, China
| | - Kai He
- School of Pharmaceutical Science, Hunan University of Medicine, Huaihua 418000, China
| | - Yong Yang
- College of Pharmacy, Hunan University of Chinese Medicine, Changsha 410000, China
| |
Collapse
|
16
|
Sibgatullina G, Al Ebrahim R, Gilizhdinova K, Tokmakova A, Malomouzh A. Differentiation of Myoblasts in Culture: Focus on Serum and Gamma-Aminobutyric Acid. Cells Tissues Organs 2023; 213:203-212. [PMID: 36871556 DOI: 10.1159/000529839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 02/20/2023] [Indexed: 03/06/2023] Open
Abstract
There are many facts about the possible role of gamma-aminobutyric acid (GABA) in the development and differentiation of cells not only in nervous but also in muscle tissue. In the present study, a primary culture of rat skeletal muscle myocytes was used to evaluate the correlation between the content of GABA in the cytoplasm and the processes of myocyte division and their fusion into myotubes. The effect of exogenous GABA on the processes of culture development was also estimated. Since the classical protocol for working with myocyte cultures involves the use of fetal bovine serum (FBS) to stimulate cell division (growth medium) and horse serum (HS) to activate the differentiation process (differentiation medium), the studies were carried out both in the medium with FBS and with HS. It was found that cells grown in medium supplemented with FBS contain more GABA compared to cultures growing in medium supplemented with HS. Addition of exogeneous GABA leads to a decrease in the number of myotubes formed in both media, while the addition of an amino acid to the medium supplemented with HS had a more pronounced inhibitory effect. Thus, we have obtained data indicating that GABA is able to participate in the early stages of skeletal muscle myogenesis by modulating the fusion process.
Collapse
Affiliation(s)
- Guzel Sibgatullina
- Laboratory of Biophysics of Synaptic Processes, Kazan Institute of Biochemistry and Biophysics, FRC Kazan Scientific Center, Russian Academy of Sciences, Kazan, Russian Federation
| | - Rahaf Al Ebrahim
- Department of Human and Animal Physiology, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russian Federation
| | - Karina Gilizhdinova
- Department of Human and Animal Physiology, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russian Federation
| | - Anna Tokmakova
- Department of Human and Animal Physiology, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russian Federation
| | - Artem Malomouzh
- Laboratory of Biophysics of Synaptic Processes, Kazan Institute of Biochemistry and Biophysics, FRC Kazan Scientific Center, Russian Academy of Sciences, Kazan, Russian Federation
| |
Collapse
|
17
|
Taurine Stimulates AMP-Activated Protein Kinase and Modulates the Skeletal Muscle Functions in Rats via the Induction of Intracellular Calcium Influx. Int J Mol Sci 2023; 24:ijms24044125. [PMID: 36835534 PMCID: PMC9962205 DOI: 10.3390/ijms24044125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 02/06/2023] [Accepted: 02/15/2023] [Indexed: 02/22/2023] Open
Abstract
Taurine (2-aminoethanesulfonic acid) is a free amino acid abundantly found in mammalian tissues. Taurine plays a role in the maintenance of skeletal muscle functions and is associated with exercise capacity. However, the mechanism underlying taurine function in skeletal muscles has not yet been elucidated. In this study, to investigate the mechanism of taurine function in the skeletal muscles, the effects of short-term administration of a relatively low dose of taurine on the skeletal muscles of Sprague-Dawley rats and the underlying mechanism of taurine function in cultured L6 myotubes were investigated. The results obtained in this study in rats and L6 cells indicate that taurine modulates the skeletal muscle function by stimulating the expression of genes and proteins associated with mitochondrial and respiratory metabolism through the activation of AMP-activated protein kinase via the calcium signaling pathway.
Collapse
|
18
|
Otten BMJ, Sthijns MMJPE, Troost FJ. A Combination of Acetate, Propionate, and Butyrate Increases Glucose Uptake in C2C12 Myotubes. Nutrients 2023; 15:nu15040946. [PMID: 36839304 PMCID: PMC9967986 DOI: 10.3390/nu15040946] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 02/09/2023] [Accepted: 02/11/2023] [Indexed: 02/16/2023] Open
Abstract
BACKGROUND Dietary fibers are subjected to saccharolytic fermentation by the gut microbiota, leading to the production of short chain fatty acids (SCFAs). SCFAs act as signaling molecules to different cells in the human body including skeletal muscle cells. The ability of SCFAs to induce multiple signaling pathways, involving nuclear erythroid 2-related factor 2 (Nrf2), may contribute to the redox balance, and thereby may be involved in glucose homeostasis. The aim of this study is to investigate whether SCFAs increase glucose uptake by upregulating the endogenous antioxidant glutathione (GSH) in C2C12 myotubes. METHODS C2C12 myotubes were exposed to 1, 5, or 20 mM of single (acetate, propionate, or butyrate) or mixtures of SCFAs for 24 h. Cytotoxicity, glucose uptake, and intracellular GSH levels were measured. RESULTS 20 mM of mixture but not separate SCFAs induced cytotoxicity. Exposure to a mixture of SCFAs at 5 mM increased glucose uptake in myotubes, while 20 mM of propionate, butyrate, and mixtures decreased glucose uptake. Exposure to single SCFAs increased GSH levels in myotubes; however, SCFAs did not prevent the menadione-induced decrease in glucose uptake in myotubes. CONCLUSIONS The effect of SCFAs on modulating glucose uptake in myotubes is not associated with the effect on endogenous GSH levels.
Collapse
|
19
|
Yin Y, Guo Q, Zhou X, Duan Y, Yang Y, Gong S, Han M, Liu Y, Yang Z, Chen Q, Li F. Role of brain-gut-muscle axis in human health and energy homeostasis. Front Nutr 2022; 9:947033. [PMID: 36276808 PMCID: PMC9582522 DOI: 10.3389/fnut.2022.947033] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 09/02/2022] [Indexed: 11/26/2022] Open
Abstract
The interrelationship between brain, gut and skeletal muscle plays a key role in energy homeostasis of the body, and is becoming a hot topic of research. Intestinal microbial metabolites, such as short-chain fatty acids (SCFAs), bile acids (BAs) and tryptophan metabolites, communicate with the central nervous system (CNS) by binding to their receptors. In fact, there is a cross-talk between the CNS and the gut. The CNS, under the stimulation of pressure, will also affect the stability of the intestinal system, including the local intestinal transport, secretion and permeability of the intestinal system. After the gastrointestinal tract collects information about food absorption, it sends signals to the central system through vagus nerve and other channels to stimulate the secretion of brain-gut peptide and produce feeding behavior, which is also an important part of maintaining energy homeostasis. Skeletal muscle has receptors for SCFAs and BAs. Therefore, intestinal microbiota can participate in skeletal muscle energy metabolism and muscle fiber conversion through their metabolites. Skeletal muscles can also communicate with the gut system during exercise. Under the stimulation of exercise, myokines secreted by skeletal muscle causes the secretion of intestinal hormones, and these hormones can act on the central system and affect food intake. The idea of the brain-gut-muscle axis is gradually being confirmed, and at present it is important for regulating energy homeostasis, which also seems to be relevant to human health. This article focuses on the interaction of intestinal microbiota, central nervous, skeletal muscle energy metabolism, and feeding behavior regulation, which will provide new insight into the diagnostic and treatment strategies for obesity, diabetes, and other metabolic diseases.
Collapse
Affiliation(s)
- Yunju Yin
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-Ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Changsha, China
| | - Qiuping Guo
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-Ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Changsha, China
| | - Xihong Zhou
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-Ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Changsha, China
| | - Yehui Duan
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-Ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Changsha, China
| | - Yuhuan Yang
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-Ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Changsha, China
| | - Saiming Gong
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-Ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Changsha, China
| | - Mengmeng Han
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-Ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Changsha, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Yating Liu
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
| | - Zhikang Yang
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
| | - Qinghua Chen
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
| | - Fengna Li
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-Ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Changsha, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
20
|
Pérez-Hernández CA, Moreno-Altamirano MMB, López-Villegas EO, Butkeviciute E, Ali M, Kronsteiner B, Dunachie SJ, Dockrell HM, Smith SG, Sánchez-García FJ. Mitochondrial Ultrastructure and Activity Are Differentially Regulated by Glycolysis-, Krebs Cycle-, and Microbiota-Derived Metabolites in Monocytes. BIOLOGY 2022; 11:biology11081132. [PMID: 36009759 PMCID: PMC9404980 DOI: 10.3390/biology11081132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 07/15/2022] [Accepted: 07/16/2022] [Indexed: 11/16/2022]
Abstract
Several intermediate metabolites harbour cell-signalling properties, thus, it is likely that specific metabolites enable the communication between neighbouring cells, as well as between host cells with the microbiota, pathogens, and tumour cells. Mitochondria, a source of intermediate metabolites, participate in a wide array of biological processes beyond that of ATP production, such as intracellular calcium homeostasis, cell signalling, apoptosis, regulation of immune responses, and host cell-microbiota crosstalk. In this regard, mitochondria's plasticity allows them to adapt their bioenergetics status to intra- and extra-cellular cues, and the mechanisms driving such plasticity are currently a matter of intensive research. Here, we addressed whether mitochondrial ultrastructure and activity are differentially shaped when human monocytes are exposed to an exogenous source of lactate (derived from glycolysis), succinate, and fumarate (Krebs cycle metabolic intermediates), or butyrate and acetate (short-chain fatty acids produced by intestinal microbiota). It has previously been shown that fumarate induces mitochondrial fusion, increases the mitochondrial membrane potential (Δψm), and reshapes the mitochondrial cristae ultrastructure. Here, we provide evidence that, in contrast to fumarate, lactate, succinate, and butyrate induce mitochondrial fission, while acetate induces mitochondrial swelling. These traits, along with mitochondrial calcium influx kinetics and glycolytic vs. mitochondrial ATP-production rates, suggest that these metabolites differentially shape mitochondrial function, paving the way for the understanding of metabolite-induced metabolic reprogramming of monocytes and its possible use for immune-response intervention.
Collapse
Affiliation(s)
- C. Angélica Pérez-Hernández
- Laboratorio de Inmunorregulación, Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City 11340, Mexico; (C.A.P.-H.); (M.M.B.M.-A.)
| | - M. Maximina Bertha Moreno-Altamirano
- Laboratorio de Inmunorregulación, Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City 11340, Mexico; (C.A.P.-H.); (M.M.B.M.-A.)
| | - Edgar O. López-Villegas
- Unidad de Microscopía, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City 11340, Mexico;
| | - Egle Butkeviciute
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London WC1E 7HT, UK; (E.B.); (H.M.D.)
| | - Mohammad Ali
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford OX1 3SY, UK; (M.A.); (B.K.); (S.J.D.)
- Oxford Centre for Global Health Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford OX3 7LG, UK
| | - Barbara Kronsteiner
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford OX1 3SY, UK; (M.A.); (B.K.); (S.J.D.)
- Oxford Centre for Global Health Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford OX3 7LG, UK
| | - Susanna J. Dunachie
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford OX1 3SY, UK; (M.A.); (B.K.); (S.J.D.)
- Oxford Centre for Global Health Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford OX3 7LG, UK
| | - Hazel M. Dockrell
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London WC1E 7HT, UK; (E.B.); (H.M.D.)
| | - Steven G. Smith
- Division of Biosciences, Brunel University London, London UB8 3PH, UK;
| | - F. Javier Sánchez-García
- Laboratorio de Inmunorregulación, Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City 11340, Mexico; (C.A.P.-H.); (M.M.B.M.-A.)
- Correspondence:
| |
Collapse
|
21
|
Martin JLA, Cartwright NM, Hutchinson AL, Robinson LE, Ma DWL, Monk JM. Differential Effects of Short-Chain Fatty Acids on L6 Myotube Inflammatory Mediator Production in Response to Lipopolysaccharide- or Palmitic Acid-Stimulation. Nutrients 2022; 14:nu14142826. [PMID: 35889783 PMCID: PMC9320465 DOI: 10.3390/nu14142826] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 06/30/2022] [Accepted: 07/05/2022] [Indexed: 11/16/2022] Open
Abstract
Short-chain fatty acids (SCFA) produced from dietary non-digestible carbohydrate fermentation have metabolic effects in skeletal muscle; however, their effect on inflammatory mediator production is unknown. In this study, L6 myotubes were cultured with individual SCFA (acetate, propionate, and butyrate) at 0.5 mM and 2.5 mM ± 10 ng/mL lipopolysaccharide (LPS) or ± 500 µM palmitic acid (PA) for 24 h. In response to LPS, only butyrate had an effect at the lower concentration (0.5 mM), whereas at the higher concentration (2.5 mM) both propionate and butyrate reduced MCP-1, MIP-1α, and RANTES secretion (p < 0.05), and only butyrate reduced IL-6 secretion and intracellular protein levels of phospho-STAT3 (p < 0.05). In response to PA, 0.5 mM butyrate reduced protein expression of phospho-NFκB p65 and the secretion of IL-6, MIP-1α, and MCP-1, whereas all three SCFA reduced RANTES secretion (p < 0.05). At the 2.5 mM SCFA concentration combined with PA stimulation, all three SCFA reduced intracellular protein expression of phospho-NFκB p65 and phospho-STAT3 and secreted protein levels of MCP-1, IL-6, and RANTES, whereas only butyrate reduced secretion of MIP-1α (p < 0.05). Thus, SCFA exhibit differential effects on inflammatory mediator expression in response to LPS and PA stimulation, which has implications for their individual impacts on inflammation-mediated skeletal muscle dysfunction.
Collapse
|
22
|
Sugimura Y, Kanda A, Sawada K, Wai KM, Tanabu A, Ozato N, Midorikawa T, Hisada T, Nakaji S, Ihara K. Association between Gut Microbiota and Body Composition in Japanese General Population: A Focus on Gut Microbiota and Skeletal Muscle. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:ijerph19127464. [PMID: 35742712 PMCID: PMC9224415 DOI: 10.3390/ijerph19127464] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 06/14/2022] [Accepted: 06/16/2022] [Indexed: 02/05/2023]
Abstract
This study aimed to investigate the gut microbial genera associated with skeletal muscle mass, using a large-scale survey from the standpoint of preventing sarcopenia. A total of 848 participants were included in the analysis. The mean (SD) ages of men (n = 353) and women (n = 495) were 50.0 (12.9) years and 50.8 (12.8) years, respectively. Body composition was assessed using appendicular skeletal muscle mass/body weight (ASM/BW), ASM, and BW. Additionally, the relationship between gut microbial genera and body composition was analyzed. The means (SD) of ASM/BW were 34.9 (2.4) % in men and 29.4 (2.9) % in women. Blautia and Bifidobacterium were positively associated with ASM/BW only in men (Blautia: β = 0.0003, Bifidobacterium: β = 0.0001). However, Blautia was negatively associated with BW (β = -0.0017). Eisenbergiella was positively associated with ASM/BW (β = 0.0209) and negatively associated with BW (β = -0.0769) only in women. Our results indicate that Blautia, Bifidobacterium and Eisenbergiella, which are positively associated with ASM/BW, might help increase skeletal muscle mass. ASM/BW may clarify the relationship between gut microbiota and skeletal muscle mass without being affected by obesity or excess body fat mass.
Collapse
Affiliation(s)
- Yoshikuni Sugimura
- Department of Innovation Center for Health Promotion, Graduate School of Medicine, Hirosaki University 5 Zaifu-cho, Hirosaki 036-8562, Japan; (Y.S.); (S.N.)
- Department of Social Medicine, Graduate School of Medicine, Hirosaki University 5 Zaifu-cho, Hirosaki 036-8562, Japan; (K.S.); (K.M.W.); (A.T.); (T.M.)
| | - Akira Kanda
- Department of Nutrition, Faculty of Health Sciences, Aomori University of Health and Welfare, 58-1 Mase, Hamadate, Aomori 030-8505, Japan;
| | - Kaori Sawada
- Department of Social Medicine, Graduate School of Medicine, Hirosaki University 5 Zaifu-cho, Hirosaki 036-8562, Japan; (K.S.); (K.M.W.); (A.T.); (T.M.)
| | - Kyi Mar Wai
- Department of Social Medicine, Graduate School of Medicine, Hirosaki University 5 Zaifu-cho, Hirosaki 036-8562, Japan; (K.S.); (K.M.W.); (A.T.); (T.M.)
| | - Asano Tanabu
- Department of Social Medicine, Graduate School of Medicine, Hirosaki University 5 Zaifu-cho, Hirosaki 036-8562, Japan; (K.S.); (K.M.W.); (A.T.); (T.M.)
| | - Naoki Ozato
- Health & Wellness Products Research Laboratories, Kao Corporation, 2-1-3 Bunka, Sumida-ku, Tokyo 131-8501, Japan;
| | - Tatsuyuki Midorikawa
- Department of Social Medicine, Graduate School of Medicine, Hirosaki University 5 Zaifu-cho, Hirosaki 036-8562, Japan; (K.S.); (K.M.W.); (A.T.); (T.M.)
- Research and Development Headquarters, Lion Corporation, Odawara 256-0811, Japan
| | - Takayoshi Hisada
- TechnoSuruga Laboratory Co., Ltd., 388-1 Nagasaki, Shimizu-ku, Shizuoka 424-0065, Japan;
| | - Shigeyuki Nakaji
- Department of Innovation Center for Health Promotion, Graduate School of Medicine, Hirosaki University 5 Zaifu-cho, Hirosaki 036-8562, Japan; (Y.S.); (S.N.)
- Department of Social Medicine, Graduate School of Medicine, Hirosaki University 5 Zaifu-cho, Hirosaki 036-8562, Japan; (K.S.); (K.M.W.); (A.T.); (T.M.)
| | - Kazushige Ihara
- Department of Social Medicine, Graduate School of Medicine, Hirosaki University 5 Zaifu-cho, Hirosaki 036-8562, Japan; (K.S.); (K.M.W.); (A.T.); (T.M.)
- Correspondence: ; Tel.: +81-172-39-5040
| |
Collapse
|
23
|
Maruta H, Abe R, Yamashita H. Effect of Long-Term Supplementation with Acetic Acid on the Skeletal Muscle of Aging Sprague Dawley Rats. Int J Mol Sci 2022; 23:ijms23094691. [PMID: 35563082 PMCID: PMC9101554 DOI: 10.3390/ijms23094691] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 04/14/2022] [Accepted: 04/21/2022] [Indexed: 12/10/2022] Open
Abstract
Mitochondrial function in skeletal muscle, which plays an essential role in oxidative capacity and physical activity, declines with aging. Acetic acid activates AMP-activated protein kinase (AMPK), which plays a key role in the regulation of whole-body energy by phosphorylating key metabolic enzymes in both biosynthetic and oxidative pathways and stimulates gene expression associated with slow-twitch fibers and mitochondria in skeletal muscle cells. In this study, we investigate whether long-term supplementation with acetic acid improves age-related changes in the skeletal muscle of aging rats in association with the activation of AMPK. Male Sprague Dawley (SD) rats were administered acetic acid orally from 37 to 56 weeks of age. Long-term supplementation with acetic acid decreased the expression of atrophy-related genes, such as atrogin-1, muscle RING-finger protein-1 (MuRF1), and transforming growth factor beta (TGF-β), activated AMPK, and affected the proliferation of mitochondria and type I fiber-related molecules in muscles. The findings suggest that acetic acid exhibits an anti-aging function in the skeletal muscles of aging rats.
Collapse
Affiliation(s)
- Hitomi Maruta
- Department of Nutritional Science, Faculty of Health and Welfare Science, Okayama Prefectural University, 111 Kuboki, Soja 719-1197, Okayama, Japan;
| | - Reina Abe
- Graduate School of Health and Welfare Science, Okayama Prefectural University, 111 Kuboki, Soja 719-1197, Okayama, Japan;
| | - Hiromi Yamashita
- Department of Nutritional Science, Faculty of Health and Welfare Science, Okayama Prefectural University, 111 Kuboki, Soja 719-1197, Okayama, Japan;
- Graduate School of Health and Welfare Science, Okayama Prefectural University, 111 Kuboki, Soja 719-1197, Okayama, Japan;
- Correspondence: ; Tel.: +81-866-94-2150
| |
Collapse
|
24
|
Zhang W, Wang W, Xu M, Xie H, Pu Z. GPR43 regulation of mitochondrial damage to alleviate inflammatory reaction in sepsis. Aging (Albany NY) 2021; 13:22588-22610. [PMID: 34584017 PMCID: PMC8507289 DOI: 10.18632/aging.203572] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 09/07/2021] [Indexed: 12/14/2022]
Abstract
Sepsis is a common critical illness in ICU and always a great difficulty in clinical treatment. GPR43 (G protein-coupled receptor 43) participates in regulating appetite and gastrointestinal peptide secretion to modulate fat decomposition and formation. However, the biological contribution of GPR43 on inflammation of sepsis has not been previously investigated. We investigated the mechanisms of GPR43 gene, which plays a possible role in distinguishing sepsis and contributes to the pathogenesis of sepsis-induced inflammatory reaction. Furthermore, we performed studies with mice induced to sepsis by Cecal Ligation and Puncture (CLP), Knockout GPR43 (GPR43-/-) mice, and Wild Type (WT) mice induced with CLP. In addition, lung tissues and cell samples were analyzed by histology, Quantitative Polymerase Chain Reaction (Q-PCR), Enzyme-linked Immunosorbent (ELISA) Assay, and western blot. GPR43 agonist could significantly reduce inflammation reactions and trigger lung injury in mice with sepsis. As for GPR43-/- mice, the risks of sepsis-induced inflammatory reactions and corresponding lung injury were promoted. On the one hand, the up-regulation of GPR43 gene reduced ROS mitochondrial damage to inhibit inflammatory reactions via the inactivation of NLRP3 Inflammasome by PPARγ/ Nox1/EBP50/ p47phox signal channel. On the other hand, the down-regulation of GPR43 promoted inflammatory reactions in vitro model through the acceleration of ROS-dependently mitochondrial damage by PPARγ/ Nox1/EBP50/ p47phox/ NLRP3 signal channel. These findings indicate that the inhibition of GPR43 as a possible important factor of sepsis may shed lights on the mechanism of sepsis-induced inflammation reaction.
Collapse
Affiliation(s)
- Weiwei Zhang
- Department of Pharmacy, Second Affiliated Hospital of Wannan Medical College, Wuhu 241001, Anhui, China
| | - Wusan Wang
- Department of Pharmacology, College of Pharmacy, Wannan Medical College, Wuhu 241002, Anhui, China
| | - Maodi Xu
- Drug Clinical Evaluation, Yijishan Hospital of Wannan Medical College, Wuhu 241001, Anhui, China
| | - Haitang Xie
- Drug Clinical Evaluation, Yijishan Hospital of Wannan Medical College, Wuhu 241001, Anhui, China
| | - Zhichen Pu
- Drug Clinical Evaluation, Yijishan Hospital of Wannan Medical College, Wuhu 241001, Anhui, China.,State Key Laboratory of Natural Medicines, Key Lab of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, Jiangsu, China
| |
Collapse
|
25
|
Navazani P, Vaseghi S, Hashemi M, Shafaati MR, Nasehi M. Effects of Treadmill Exercise on the Expression Level of BAX, BAD, BCL-2, BCL-XL, TFAM, and PGC-1α in the Hippocampus of Thimerosal-Treated Rats. Neurotox Res 2021; 39:1274-1284. [PMID: 33939098 DOI: 10.1007/s12640-021-00370-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 04/10/2021] [Accepted: 04/26/2021] [Indexed: 01/11/2023]
Abstract
Thimerosal (THIM) induces neurotoxic changes including neuronal death and releases apoptosis inducing factors from mitochondria to cytosol. THIM alters the expression level of factors involved in apoptosis. On the other hand, the anti-apoptotic effects of exercise have been reported. In this study, we aimed to discover the effect of three protocols of treadmill exercise on the expression level of mitochondrial transcription factor A (TFAM), peroxisome proliferator-activated receptor γ coactivator-1α (PGC-1α), BCL-2-associated death (BAD), BCL-2-associated X (BAX), BCL-XL, and BCL-2 (a pro-survival BCL-2 protein) in the hippocampus of control and THIM-exposed rats. Male Wistar rats were used in this research. Real-time PCR was applied to assess genes expression. The results showed that THIM increased the expression of pro-apoptotic factors (BAD and BAX), decreased the expression of anti-apoptotic factors (BCL-2 and BCL-XL), and decreased the expression of factors involved in mitochondrial biogenesis (TFAM and PGC-1α). Treadmill exercise protocols reversed the effect of THIM on all genes. In addition, treadmill exercise protocols decreased the expression of BAD and BAX, increased the expression of BCL-2, and increased the expression of TFAM and PGC-1α in control rats. In conclusion, THIM induced a pro-apoptotic effect and disturbed mitochondrial biogenesis and stability, whereas treadmill exercise reversed these effects.
Collapse
Affiliation(s)
- Pouria Navazani
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Salar Vaseghi
- Cognitive and Neuroscience Research Center (CNRC), Amir-Almomenin Hospital, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.,Department of Cognitive Neuroscience, Institute for Cognitive Science Studies (ICSS), Tehran, Iran
| | - Mehrdad Hashemi
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Mohammad-Reza Shafaati
- Department of Cellular and Molecular Biology, Faculty of Basic Sciences, Hamadan Branch, Islamic Azad University, Hamadan, Iran
| | - Mohammad Nasehi
- Cognitive and Neuroscience Research Center (CNRC), Amir-Almomenin Hospital, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| |
Collapse
|
26
|
Carretta MD, Quiroga J, López R, Hidalgo MA, Burgos RA. Participation of Short-Chain Fatty Acids and Their Receptors in Gut Inflammation and Colon Cancer. Front Physiol 2021; 12:662739. [PMID: 33897470 PMCID: PMC8060628 DOI: 10.3389/fphys.2021.662739] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 03/09/2021] [Indexed: 02/06/2023] Open
Abstract
Short-chain fatty acids (SCFAs) are the main metabolites produced by the bacterial fermentation of dietary fiber, and they play a critical role in the maintenance of intestinal health. SCFAs are also essential for modulating different processes, and they have anti-inflammatory properties and immunomodulatory effects. As the inflammatory process predisposes the development of cancer and promotes all stages of tumorigenesis, an antitumor effect has also been associated with SCFAs. This is strongly supported by epidemiological studies showing that a diet rich in fiber is linked to a reduced risk of colon cancer and has significant clinical benefits in patients with inflammatory bowel disease (IBD). SCFAs may signal through the metabolite-sensing G protein-coupled receptors free fatty acid receptor 3 [FFAR3 or G protein-coupled receptor 41 (GPR41)], FFAR2 (GPR43), and GPR109A (also known as hydroxycarboxylic acid receptor 2 or HCAR2) expressed in the gut epithelium and immune cells. This review summarizes the existing knowledge regarding the SCFA-mediated suppression of inflammation and carcinogenesis in IBD and colon cancer.
Collapse
Affiliation(s)
- María Daniella Carretta
- Laboratory of Inflammation Pharmacology, Faculty of Veterinary Science, Institute of Pharmacology and Morphophysiology, Universidad Austral de Chile, Valdivia, Chile
| | | | | | | | - Rafael Agustín Burgos
- Laboratory of Inflammation Pharmacology, Faculty of Veterinary Science, Institute of Pharmacology and Morphophysiology, Universidad Austral de Chile, Valdivia, Chile
| |
Collapse
|
27
|
Ma Y, Maruta H, Sun B, Wang C, Isono C, Yamashita H. Effects of long-term taurine supplementation on age-related changes in skeletal muscle function of Sprague-Dawley rats. Amino Acids 2021; 53:159-170. [PMID: 33398526 DOI: 10.1007/s00726-020-02934-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 12/19/2020] [Indexed: 12/16/2022]
Abstract
Taurine (2-aminoethanesulfonic acid) is a free amino acid found abundantly in mammalian tissues. Increasing evidence suggests that taurine plays a role in the maintenance of skeletal muscle function and increase of exercise capacity. Most energy drinks contain this amino acid; however, there is insufficient research on the effects of long-term, low-dose supplementation of taurine. In this study, we investigated the effects of long-term administration of taurine at low doses on aging in rodents. In Experiment 1, we examined age-related changes in aging Sprague-Dawley (SD) rats (32-92 weeks old) that O2 consumption and spontaneous activity decreased significantly with aging. In Experiment 2, we examined the effects of long-term (21-week) administration of taurine on healthy aging SD rats. SD rats were stabilized for 32-34 weeks and divided into three groups, administrated water (control), 0.5% taurine (25 mg/kg body weight (BW)/day), or 1% taurine (50 mg/kg BW/day) from age 34 to 56 weeks (5 days/week, 5 mL/kg BW). Our findings suggest that long-term administration of taurine at relatively low dose could attenuate the age-related decline in O2 consumption and spontaneous locomotor activity. Upon intestinal absorption, taurine might modulate age-related changes in respiratory metabolism and skeletal muscle function via peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α), succinate dehydrogenase (SDH), cytochrome c (Cycs), myocyte enhancer factor 2A (MEF2A), glucose transporter 4 (GLUT4), and myoglobin, which are regulated by the activation of AMP-activated protein kinase (AMPK). This article examines the mechanism underlying the effects of taurine on age-related changes, which may have potential clinical implications.
Collapse
Affiliation(s)
- Yun Ma
- Graduate School of Health and Welfare Science, Okayama Prefectural University, Soja, Okayama, 719-1197, Japan
| | - Hitomi Maruta
- Department of Nutritional Science, Okayama Prefectural University, Soja, Okayama, 719-1197, Japan
| | - Baojun Sun
- Graduate School of Health and Welfare Science, Okayama Prefectural University, Soja, Okayama, 719-1197, Japan
| | - Chengduo Wang
- Graduate School of Health and Welfare Science, Okayama Prefectural University, Soja, Okayama, 719-1197, Japan
| | - Chiaki Isono
- Graduate School of Health and Welfare Science, Okayama Prefectural University, Soja, Okayama, 719-1197, Japan
| | - Hiromi Yamashita
- Graduate School of Health and Welfare Science, Okayama Prefectural University, Soja, Okayama, 719-1197, Japan. .,Department of Nutritional Science, Okayama Prefectural University, Soja, Okayama, 719-1197, Japan.
| |
Collapse
|